Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Int J Cancer ; 151(1): 107-119, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35179790

ABSTRACT

Lynch Syndrome (LS) is an autosomal dominant genetic condition that causes a high risk of colorectal cancer. The hallmark of LS is genetic instability as a result of mismatch repair (MMR) deficiency, particularly in repetitive low complexity regions called microsatellites (MS). MLH1-/- mice deficient in MMR are prone to developing tumors in the colon, upon oral administration of dextran sodium sulfate (DSS), at a rate of more than 70%. Using this LS mouse model, we found a novel tumor neo-antigen from a deletion mutation of the coding MS in the SENP6 gene that prevented tumorigenesis or hindered tumor growth rate in immunized mice. This was accomplished via high throughput exome sequencing of DSS-induced colorectal tumors in the MLH1-/- mice and predicting the most highly immunogenic mutant gene products processed and presented as antigens in C57BL/6 MHC-I molecules. Throughout our study, we were able to prove the validity of the vaccine by analyzing the colorectal tumors in immunized DSS-treated mice using either our epitope, called Sp6D1, or an unrelated peptide as a negative control. Tumors developed in this context were found to be antigenic and Sp6D1-specific CD8+ tumor infiltrating lymphocytes were detected by flow cytometry and cytotoxic T lymphocytes (CTL) killing assays. Additionally, immunohistochemistry showed that tumor-adjacent tertiary lymphoid organs were a potentially significant source of CD8+ lymphocytes. Altogether, our results indicate that there may be a protective effect to patients carrying LS mutations through the induction of a peptide-specific CTL response from the use of neoepitope vaccination.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis , Colorectal Neoplasms , Vaccines , Animals , Antigens, Neoplasm/genetics , Brain Neoplasms , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/prevention & control , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/prevention & control , Cysteine Endopeptidases/genetics , DNA Mismatch Repair/genetics , Humans , Mice , Mice, Inbred C57BL , Microsatellite Instability , MutL Protein Homolog 1/genetics , Neoplastic Syndromes, Hereditary
2.
Eur J Immunol ; 51(6): 1505-1518, 2021 06.
Article in English | MEDLINE | ID: mdl-33835499

ABSTRACT

A T-cell receptor (TCR) with optimal avidity to a tumor antigen can be used to redirect T cells to eradicate cancer cells via adoptive cell transfer. Cancer testis antigens (CTAs) are attractive targets because they are expressed in the testis, which is immune-privileged, and in the tumor. However, CTAs are self-antigens and natural TCRs to CTAs have low affinity/avidity due to central tolerance. We previously described a method of directed evolution of TCR avidity using somatic hypermutation. In this study, we made several improvements to this method and enhanced the avidity of the hT27 TCR, which is specific for the cancer testis antigen HLA-A2-MAGE-A1278-286 . We identified eight point mutations with varying degrees of improved avidity. Human T cells transduced with TCRs containing these mutations displayed enhanced tetramer binding, IFN-γ and IL2 production, and cytotoxicity. Most of the mutations have retained specificity, except for one mutant with extremely high avidity. We demonstrate that somatic hypermutation is capable of optimizing avidity of clinically relevant TCRs for immunotherapy.


Subject(s)
Cancer Vaccines/immunology , Immunotherapy, Adoptive/methods , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , Cells, Cultured , Central Tolerance , Cytotoxicity, Immunologic , HLA-A2 Antigen/metabolism , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Point Mutation/genetics , Protein Binding , Receptors, Antigen, T-Cell/metabolism , Somatic Hypermutation, Immunoglobulin , T-Lymphocytes/transplantation
3.
Int J Cancer ; 145(10): 2816-2826, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31381134

ABSTRACT

Adoptive transfer of T cells that have been genetically modified to express an antitumor T-cell receptor (TCR) is a potent immunotherapy, but only if TCR avidity is sufficiently high. Endogenous TCRs specific to shared (self) tumor-associated antigens (TAAs) have low affinity due to central tolerance. Therefore, for effective therapy, anti-TAA TCRs with higher and optimal avidity must be generated. Here, we describe a new in vitro system for directed evolution of TCR avidity using somatic hypermutation (SHM), a mechanism used in nature by B cells for antibody optimization. We identified 44 point mutations to the Pmel-1 TCR, specific for the H-2Db -gp10025-33 melanoma antigen. Primary T cells transduced with TCRs containing two or three of these mutations had enhanced activity in vitro. Furthermore, the triple-mutant TCR improved in vivo therapy of tumor-bearing mice, which exhibited improved survival, smaller tumors and delayed or no relapse. TCR avidity maturation by SHM may be an effective strategy to improve cancer immunotherapy.


Subject(s)
Directed Molecular Evolution/methods , Melanoma, Experimental/therapy , Receptors, Antigen, T-Cell/genetics , Skin Neoplasms/therapy , gp100 Melanoma Antigen/immunology , Animals , Cell Line, Tumor , HEK293 Cells , Histocompatibility Antigen H-2D , Humans , Immunotherapy, Adoptive/methods , Melanoma, Experimental/immunology , Mice , Mice, Transgenic , Mutagenesis, Site-Directed/methods , Peptides/immunology , Point Mutation , Proof of Concept Study , Receptors, Antigen, T-Cell/immunology , Skin Neoplasms/immunology
4.
Int J Cancer ; 144(4): 909-921, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30106470

ABSTRACT

For many years, clinicians and scientists attempt to develop methods to stimulate the immune system to target malignant cells. Recent data suggest that effective cancer vaccination requires combination immunotherapies to overcome tumor immune evasion. Through presentation of both MHC-I and II molecules, DCs-based vaccine platforms are effective in generating detectable CD4 and CD8 T cell responses against tumor-associated antigens. Several platforms include DC transfection with mRNA of the desired tumor antigen. These DCs are then delivered to the host and elicit an immune response against the antigen of interest. We have recently established an mRNA genetic platform which induced specific CD8+ cytotoxic T cell response by DC vaccination against melanoma. In our study, an MHC-II mRNA DCs vaccine platform was developed to activate CD4+ T cells and to enhance the anti-tumor response. The invariant chain (Ii) was modified and the semi-peptide CLIP was replaced with an MHC-II binding peptide sequences of melanoma antigens. These chimeric MHC-II constructs are presented by DCs and induce proliferation of tumor specific CD4+ T cells. When administered in combination with the MHC-I platform into tumor bearing mice, these constructs were able to inhibit tumor growth, and improve mouse survival. Deciphering the immunological mechanism of action, we observed an efficient CTLs killing in addition to higher levels of Th1 and Th2 subsets in the groups immunized with a combination of the MHC-I and MHC-II constructs. These universal constructs can be applied in multiple combinations and offer an attractive opportunity to improve cancer treatment.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/immunology , Cancer Vaccines/immunology , Histocompatibility Antigens Class II/immunology , Melanoma, Experimental/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Progression , Immunotherapy/methods , Melanoma, Experimental/genetics , Melanoma, Experimental/therapy , Mice, Inbred C57BL , Mice, Transgenic , Survival Analysis , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
5.
Oncoimmunology ; 7(7): e1445457, 2018.
Article in English | MEDLINE | ID: mdl-29900058

ABSTRACT

Despite melanoma immunogenicity and remarkable therapeutic effects of negative immune checkpoint inhibitors, a significant fraction of patients does not respond to current treatments. This could be due to limitations in tumor immunogenicity and profound immunosuppression in the melanoma microenvironment. Moreover, insufficient tumor antigen processing and presentation by dendritic cells (DC) may hamper the development of tumor-specific T cells. Using two genetically engineered mouse melanoma models (RET and BRAFV600E transgenic mice), in which checkpoint inhibitor therapy alone is not efficacious, we performed proof-of-concept studies with an improved, multivalent DC vaccination strategy based on our recently developed genetic mRNA cancer vaccines. The in vivo expression of multiple chimeric MHC class I receptors allows a simultaneous presentation of several melanoma-associated shared antigens tyrosinase related protein (TRP)-1, tyrosinase, human glycoprotein 100 and TRP-2. The DC vaccine induced a significantly improved survival in both transgenic mouse models. Vaccinated melanoma-bearing mice displayed an increased CD8 T cell reactivity indicated by a higher IFN-γ production and an upregulation of activation marker expression along with an attenuated immunosuppressive pattern of myeloid-derived suppressor cells (MDSC) and regulatory T cells (Treg). The combination of DC vaccination with ultra-low doses of paclitaxel or anti-PD-1 antibodies resulted in further prolongation of mouse survival associated with a stronger reduction of MDSC and Treg immunosuppressive phenotype. Our data suggest that an improved multivalent DC vaccine based on shared tumor antigens induces potent anti-tumor effects and could be combined with checkpoint inhibitors or targeting immunosuppressive cells to further improve their therapeutic efficiency.

6.
Immunol Cell Biol ; 96(3): 284-297, 2018 03.
Article in English | MEDLINE | ID: mdl-29356071

ABSTRACT

Inflammation plays pivotal roles in different stages of tumor development. Screening for predisposing genetic abnormalities and understanding the roles these genes play in the crosstalk between immune and cancer cells will provide new targets for cancer therapy and prevention. The interferon inducible transmembrane (IFITM) genes are involved in pathogenesis of the gastro-intestinal tract. We aimed at delineating the role of IFITM3 in colonic epithelial homeostasis, inflammation and colitis-associated tumorigenesis using IFITM3-deficient mice. Chemical induction of colitis in IFITM3-deficient mice results in significantly increased clinical signs of inflammation and induction of invasive tumorigenesis. Bone marrow transplantation showed that cells of the hematopoietic system are responsible for colitis deterioration. In these mice, impaired cytokine expression skewed inflammatory response toward pathogenic Th17 with reduced expression of the anti-inflammatory cytokine IL10 during the recovery phase. Intriguingly, mice lacking the entire IFITM locus developed spontaneous chronic colitis from the age of 14 weeks. Sequencing the 16S rRNA of naïve mice lacking IFITM3 gene, or the entire locus containing five IFITM genes, revealed these mice had significant bacterial differences from their wild-type littermates. Our novel results provide strong evidence for the essential role of IFITM genes in ameliorating colitis and colitis-associated tumorigenesis.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/pathology , Colitis/immunology , Colitis/microbiology , Immunity , Inflammation/genetics , Membrane Proteins/genetics , Microbiota , Animals , Colitis/genetics , Colitis/pathology , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Disease Progression , Disease Susceptibility , Hematopoiesis , Immunity/genetics , Membrane Proteins/deficiency , Mice, Inbred C57BL , Mice, Knockout , Microbiota/genetics , Myeloid Cells/pathology
7.
Oncoimmunology ; 5(6): e1160183, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27471629

ABSTRACT

Malignant melanoma is characterized by a rapid progression, metastasis to distant organs and resistance to chemo and radiotherapy. Although melanoma is capable of eliciting an immune response, the disease progresses and the overall results of immunotherapeutic clinical studies are not satisfactory. Recently, we have developed a novel genetic platform for improving an induction of peptide-specific CD8(+) T cells by dendritic cell (DC) based on membrane-anchored ß2-microglobulin (ß2m) linked to a selected antigenic peptide at the N-terminus and to the cytosolic domain of TLR4 at the C-terminus. In vitro transcribed mRNA transfection of antigen-presenting cells (APCs) resulted in an efficient coupling of peptide presentation and cell activation. In this research, we utilize the chimeric platform to induce an immune response in ret transgenic mice that spontaneously develop malignant skin melanoma and to examine its effect on the overall survival of tumor-bearing mice. Following immunization with chimeric construct system, we observe a significantly prolonged survival of tumor-bearing mice as compared to the control group. Moreover, we see elevations in the frequency of CD62L(hi)CD44(hi) central and CD62L(lo)CD44(hi) effector memory CD8(+) T-cell subsets. Importantly, we do not observe any changes in frequencies of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) in the vaccinated groups. Our data suggest that this novel vaccination approach could be efficiently applied for the immunotherapy of malignant melanoma.

8.
Mol Ther ; 23(8): 1391-1400, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25997427

ABSTRACT

Recently, we have developed a novel genetic platform for improving dendritic cell (DC) induction of peptide-specific CD8 T cells, based on membrane-anchored ß2-microglobulin (ß2m) linked to a selected antigenic peptide at its N-terminus and to the cytosolic domain of toll-like receptor (TLR)4 C-terminally. In vitro transcribed mRNA transfection of antigen presenting cells resulted in polypeptides that efficiently coupled peptide presentation to cellular activation. In the present study, we evaluated the immunogenicity of such constructs in mRNA-transfected immature murine bone marrow-derived DCs. We show that the encoded peptide ß2m-TLR4 products were expressed at the cell surface up to 72 hours and stimulated the maturation of DCs. In vivo, these DCs prompted efficient peptide-specific T-cell activation and target cell killing which were superior to those induced by peptide-loaded, LPS-stimulated DCs. This superiority was also evident in the ability to protect mice from tumor progression following the administration of B16F10.9 melanoma cells and to inhibit the development of pre-established B16F10.9 tumors. Our results provide evidence that the products of two recombinant genes encoding for peptide-hß2m-TLR4 and peptide-hß2m-K(b) expressed from exogenous mRNA can cooperate to couple Major Histocompatibility Complex (MHC-I) peptide presentation to TLR-mediated signaling, offering a safe, economical and highly versatile genetic platform for a novel category of CTL-inducing vaccines.


Subject(s)
Dendritic Cells/cytology , Histocompatibility Antigens Class I/metabolism , Peptides/chemistry , RNA, Messenger/metabolism , Toll-Like Receptor 4/metabolism , beta 2-Microglobulin/metabolism , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/cytology , Antigens, Neoplasm/chemistry , Antineoplastic Agents/chemistry , Bone Marrow Cells/cytology , Cancer Vaccines/immunology , Cell Membrane/metabolism , Cloning, Molecular , Culture Media , Cytosol/metabolism , Female , Humans , Lipopolysaccharides/chemistry , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Protein Structure, Tertiary , Signal Transduction
9.
Cancer Immunol Immunother ; 63(4): 369-80, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24452202

ABSTRACT

Cryoablation is a low-invasive surgical procedure for management of malignant tumors. Tissue destruction is obtained by repeated deep freezing and thawing and results in coagulative necrosis and in apoptosis. This procedure induces the release of tumor-associated antigens and proinflammatory factors into the microenvironment. Local administration of immature dendritic cells (DCs) potentiates the immune response induced by cryoablation. To further augment the induction of long-lasting antitumor immunity, we investigated the clinical value of combining cryoimmunotherapy consisting of cryoablation and inoculation of immature DCs with administration of the immune adjuvant, CpG oligodeoxynucleotides. Injection of the murine Lewis lung carcinoma, D122-luc-5.5 that expresses the luciferase gene, results in spontaneous metastases, which can be easily monitored in vivo. The local tumor was treated by the combined treatment. The clinical outcome was assessed by monitoring tumor growth, metastasis in distant organs, overall survival, and protection from tumor recurrence. The nature of the induced T cell responses was analyzed. Combined cryoimmunotherapy results in reduced tumor growth, low metastasis and significantly prolonged survival. Moreover, this treatment induces antitumor memory that protected mice from rechallenge. The underlying suggested mechanisms are the generation of tumor-specific type 1 T cell responses, subsequent induction of cytotoxic T lymphocytes, and generation of systemic memory. Our data highlight the combined cryoimmunotherapy as a novel antitumor vaccine with promising preclinical results. Adjustment of this technique into practice will provide the therapeutic benefits of both, ablation of the primary tumor and induction of robust antitumor and antimetastatic immunity.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Carcinoma, Lewis Lung/therapy , CpG Islands , Cryosurgery/methods , Dendritic Cells/immunology , Immunotherapy/methods , Lung Neoplasms/therapy , Oligodeoxyribonucleotides/therapeutic use , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/secondary , Carcinoma, Lewis Lung/surgery , Cells, Cultured , Combined Modality Therapy , Dendritic Cells/transplantation , Foot , Immunologic Memory , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lung Neoplasms/surgery , Male , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Oligodeoxyribonucleotides/administration & dosage , Recurrence , Specific Pathogen-Free Organisms , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology
10.
Ann N Y Acad Sci ; 1283: 87-90, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23387369

ABSTRACT

Cytotoxic T lymphocytes (CTLs) are the major effector arm of the immune system against tumors. Many tumor-associated antigens (TAAs), known today as potential rejection antigens, were identified by their ability to induce CTL responses. CTLs utilize their clonotypic T cell receptor (TCR) to recognize short antigenic peptides presented on major histocompatibility complex (MHC)-I proteins. These consist of a membrane-attached α heavy chain, which forms the peptide binding pocket, and a noncovalently associated ß2m light chain, not anchored to the cell membrane. CTL activation requires that antigenic peptides be presented initially on professional antigen presenting cells (APCs), primarily dendritic cells (DCs). Autologous DCs are a powerful tool for the induction of antitumor responses and are thus widely explored as vehicles for cancer vaccines. Although encouraging evidence for the induction of tumor-specific CTLs by ex vivo-manipulated DCs came from numerous animal studies, reproducible objective clinical response in human trials is yet to be demonstrated.


Subject(s)
Cancer Vaccines/genetics , Cancer Vaccines/immunology , beta 2-Microglobulin/genetics , beta 2-Microglobulin/metabolism , Animals , Cancer Vaccines/therapeutic use , Drug Design , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/therapeutic use , Mice , Protein Engineering , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use
11.
PLoS One ; 8(2): e55583, 2013.
Article in English | MEDLINE | ID: mdl-23383339

ABSTRACT

Identification and quantification of immunogenic peptides and tumor-derived epitopes presented on MHC-I molecules are essential for basic studies and vaccines generation. Although lymphocytes derived from transgenic mice can serve as sensitive detectors of processes of antigen presentation and recognition, they are not always available. The use of cell lines might be extremely useful. In this study, we generated a lacZ inducible CD8⁺ hybridoma (BUSA14) capable of recognizing both human and mouse gp10025₋33 melanoma antigens presented on dendritic and tumor cell lines. This hybridoma expresses a variety of membranal T cell markers and secretes IL-2 and TNFα. Thus, BUSA14 offers a quantifiable, cheap and straightforward tool for studying peptide presentation by MHC-I molecules on the cell surface.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Histocompatibility Antigens Class I/metabolism , Hybridomas/metabolism , gp100 Melanoma Antigen/metabolism , Animals , Cell Line, Tumor , Dendritic Cells/metabolism , Flow Cytometry , Humans , Hybridomas/cytology , Interleukin-2/metabolism , Lac Operon , Mice , Tumor Necrosis Factor-alpha/metabolism
12.
Int Immunol ; 23(7): 453-61, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21652516

ABSTRACT

Priming of naive CD8 T cells by dendritic cells (DCs) entails both effective antigen presentation on MHC class I products and co-stimulatory signaling. Their optimal coupling is a major goal in the development of CTL-inducing vaccines. We recently reported that a membranal derivative of the invariant MHC-I light chain, ß(2)-microglobulin (ß(2)m), markedly stabilizes MHC-I molecules and can serve as a universal platform for exceptional presentation of genetically linked peptides. To test whether it is possible to equip the resulting MHC-I complexes with an inherent ability to activate antigen-presenting cells, we engrafted the intracellular Toll/IL-1 receptor domain of mouse Toll-like receptor (TLR) 4 or TLR2 onto the peptide-ß(2)m scaffold. We evaluated the level of peptide presentation and status of cell activation conferred by such constructs in stably transfected mouse RAW264.7 macrophages and mRNA-transfected mouse DC2.4 DCs. We show that the encoded peptide-ß(2)m-TLR polypeptides are expressed at the cell surface, pair with endogenous heavy chains, stabilize MHC-I products, prompt efficient peptide-specific T-cell recognition and confer a constitutively activated phenotype on the transfected cells, as judged by the up-regulation of pro-inflammatory genes and surface co-stimulatory molecules. Our results provide evidence that the product of a single recombinant gene can couple MHC peptide presentation to TLR-mediated signaling and offer a safe, economical and highly versatile modality for a novel category of genetic CTL-inducing vaccines.


Subject(s)
Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , H-2 Antigens/immunology , Peptides/immunology , Animals , Genetic Vectors/genetics , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptides/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Transfection , beta 2-Microglobulin/genetics , beta 2-Microglobulin/immunology
13.
Int J Cancer ; 125(12): 2810-9, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19544527

ABSTRACT

The human 1-8 interferon inducible gene family consists of at least 3 functional genes; 9-27, 1-8D and 1-8U, which are all linked on an 18-kb fragment of chromosome 11 and are highly homologous. It has recently been shown by us and others that the 1-8D gene is overexpressed in colon carcinoma. Here, we show, by sequence comparison of the 1-8D in pairs of tumor/normal colon tissues, the existence of 6 different alleles, containing single-nucleotide polymorphisms with no mutations. Transformation assays revealed a possible role for the 1-8D gene as a transformation inhibitor. Further, transient expression of the human 1-8D gene in multiple mammalian cell lines showed accumulation of cells in the G1 phase followed by elevation in the subG1 phase. SubG1 elevation was confirmed as apoptosis by Annexin-V binding assays and transferase-mediated dUTP nick end labeling assays. Moreover, knock-down of 1-8D provided partial protection from Etoposide and UV-induced apoptosis. The induction of apoptosis by 1-8D is dependent on caspase activities but not on p53 expression. Although 1-8D induces apoptosis independently of p53, p53 expression downregulates 1-8D protein expression. Our data suggest a role for the 1-8D gene as a novel pro-apoptotic gene that will provide new insights into the regulated cellular pathways to death.


Subject(s)
Apoptosis/genetics , Cell Transformation, Neoplastic , Membrane Proteins/genetics , Polymorphism, Single Nucleotide/genetics , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Animals , Annexin A5/metabolism , Blotting, Western , Case-Control Studies , Caspases/metabolism , Cell Line, Tumor , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , G1 Phase , Humans , Membrane Proteins/antagonists & inhibitors , Mice , Molecular Sequence Data , Mutation/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Tumor Suppressor Protein p53/genetics
14.
Cancer Res ; 68(9): 3450-7, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18451173

ABSTRACT

Cytotoxic T cells (CTL) play a major role in tumor rejection. Expansion of CTLs, either by immunization or adoptive transfer, is a prominent goal in current immunotherapy. The antigen-specific nature of these expansion processes inevitably initiates a clonotypic attack on the tumor. By injecting an Ovalbumin-expressing melanoma into OT-I mice, in which >90% of CTLs recognize an Ovalbumin peptide, we show that an increased number of tumor-specific CTLs causes emergence of escape variants. We show that these escape variants are a result of antigen silencing via a yet undetermined epigenetic mechanism, which occurs frequently and is spontaneously reversible. We further show that an increase in the time of tumor onset in OT-I compared with C57BL/6J is a result of immune selection.


Subject(s)
Lymphocytes, Tumor-Infiltrating/pathology , Melanoma, Experimental/immunology , T-Lymphocytes/pathology , Tumor Escape/immunology , Adoptive Transfer , Animals , Antigens, Neoplasm/genetics , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Proliferation , Gene Silencing/physiology , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Proteins/genetics , Ovalbumin/genetics , RNA Editing/immunology , RNA Editing/physiology
15.
Cancer Immunol Immunother ; 56(2): 217-26, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16738849

ABSTRACT

Conventional treatment of recurrent and metastasized prostate cancer (CaP) remains inadequate; this fact mandates development of alternative therapeutic modalities, such as specific active or passive immunotherapy. Previously, we reported the identification of a novel highly immunogenic HLA-A*0201-restricted Prostatic Acid Phosphatase-derived peptide (PAP-3) by a two-step in vivo screening in an HLA-transgenic (HHD) mouse system. In the present study we aimed at elucidating the efficiency of PAP-3-based vaccine upon active antitumor immunization. To this end we established preventive and therapeutic carcinoma models in HHD mice. The 3LL murine Lewis lung carcinoma clone D122 transduced to express HLA-A*0201 and PAP served as a platform for these models. The HLA-A*0201-PAP-3 complex specific recombinant single chain scFV-PAP-3 antibodies were generated and used to confirm an endogenous PAP processing resulting in PAP-3 presentation by HLA-A*0201. PAP-3 based vaccines significantly decreased tumor incidence in a preventive immunization setting. Therapeutic vaccination of HHD mice with PAP-3 led to rejection of early established tumors and to increase of mouse survival. These results strongly support a therapeutic relevance of the identified CTL epitope upon active antitumor immunization. The newly established carcinoma model presented herein might be a useful tool for cancer vaccine design and optimization.


Subject(s)
Cancer Vaccines/immunology , Immunotherapy/methods , Lymphokines/immunology , Prostatic Neoplasms/immunology , Protein Tyrosine Phosphatases/immunology , Sialoglycoproteins/immunology , Acid Phosphatase , Animals , Cancer Vaccines/therapeutic use , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/therapy , Epitopes, T-Lymphocyte/immunology , HLA-A Antigens/immunology , HLA-A2 Antigen , Humans , Lymphokines/therapeutic use , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Polymerase Chain Reaction , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/therapeutic use , Sialoglycoproteins/therapeutic use
16.
J Immunol ; 176(1): 217-24, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16365413

ABSTRACT

Level and persistence of antigenic peptides presented by APCs on MHC class I (MHC-I) molecules influence the magnitude and quality of the ensuing CTL response. We recently demonstrated the unique immunological properties conferred on APCs by expressing beta2-microglobulin (beta2m) as an integral membrane protein. In this study, we explored membrane-anchored beta2m as a platform for cancer vaccines using as a model MO5, an OVA-expressing mouse B16 melanoma. We expressed in mouse RMA-S cells two H-2Kb binding peptides from MO5, OVA257-264, and TRP-2181-188, each genetically fused with the N terminus of membranal beta2m via a short linker. Specific Ab staining and T cell hybridoma activation confirmed that OVA257-264 was properly situated in the MHC-I binding groove. In vivo, transfectants expressing both peptides elicited stronger CTLs and conferred better protection against MO5 than peptide-saturated RMA-S cells. Cells expressing OVA257-264/beta2m were significantly superior to OVA257-264-charged cells in their ability to inhibit the growth of pre-established MO5 tumors. Our results highlight the immunotherapeutic potential of membranal beta2m as a universal scaffold for optimizing Ag presentation by MHC-I molecules.


Subject(s)
Cancer Vaccines/immunology , Epitopes, T-Lymphocyte/immunology , Melanoma, Experimental/immunology , T-Lymphocytes, Cytotoxic/immunology , beta 2-Microglobulin/immunology , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Cell Membrane/immunology , Cell Membrane/metabolism , Flow Cytometry , H-2 Antigens/chemistry , H-2 Antigens/genetics , H-2 Antigens/immunology , Mice , Transfection , beta 2-Microglobulin/chemistry , beta 2-Microglobulin/genetics
17.
Cancer Res ; 65(14): 6435-42, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-16024648

ABSTRACT

Specific immunotherapy of prostate cancer may be an alternative or be complementary to other approaches for treatment of recurrent or metastasized disease. This study aims at identifying and characterizing prostate cancer-associated peptides capable of eliciting specific CTL responses in vivo. Evaluation of peptide-induced CTL activity in vitro was done following immunization of HLA-A2 transgenic (HHD) mice. An in vivo tumor rejection was tested by adoptive transfer of HHD immune lymphocytes to nude mice bearing human tumors. To confirm the existence of peptide-specific CTL precursors in human, lymphocytes from healthy and prostate cancer individuals were stimulated in vitro in the presence of these peptides and CTL activities were assayed. Two novel immunogenic peptides derived from overexpressed prostate antigens, prostatic acid phosphatase (PAP) and six-transmembrane epithelial antigen of prostate (STEAP), were identified; these peptides were designated PAP-3 and STEAP-3. Peptide-specific CTLs lysed HLA-A2.1+ LNCaP cells and inhibited tumor growth on adoptive immunotherapy. Furthermore, peptide-primed human lymphocytes derived from healthy and prostate cancer individuals lysed peptide-pulsed T2 cells and HLA-A2.1+ LNCaP cells. Based on the results presented herein, PAP-3 and STEAP-3 are naturally processed CTL epitopes possessing anti-prostate cancer reactivity in vivo and therefore may constitute vaccine candidates to be investigated in clinical trials.


Subject(s)
Antigens, Neoplasm/immunology , Epitopes, T-Lymphocyte/immunology , Immunotherapy, Adoptive/methods , Peptide Fragments/immunology , Prostatic Neoplasms/immunology , Protein Tyrosine Phosphatases/immunology , T-Lymphocytes, Cytotoxic/immunology , Acid Phosphatase , Amino Acid Sequence , Animals , Cell Line, Tumor , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Humans , Male , Mice , Mice, Knockout , Mice, Nude , Mice, Transgenic , Oxidoreductases , Prostatic Neoplasms/therapy , Xenograft Model Antitumor Assays
18.
Clin Cancer Res ; 11(13): 4955-61, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-16000595

ABSTRACT

PURPOSE: Cryotherapy of localized prostate, renal, and hepatic primary tumors and metastases is considered a minimally invasive treatment demonstrating a low complication rate in comparison with conventional surgery. The main drawback of cryotherapy is that it has no systemic effect on distant metastases. We investigated whether intratumoral injections of dendritic cells following cryotherapy of local tumors (cryoimmunotherapy) provides an improved approach to cancer treatment, combining local tumor destruction and systemic anticancer immunity. EXPERIMENTAL DESIGNS: The 3LL murine Lewis lung carcinoma clone D122 and the ovalbumin-transfected B16 melanoma clone MO5 served as models for spontaneous metastasis. The antimetastatic effect of cryoimmunotherapy was assessed in the lung carcinoma model by monitoring mouse survival, lung weight, and induction of tumor-specific CTLs. The mechanism of cryoimmunotherapy was elucidated in the melanoma model using adoptive transfer of T cell receptor transgenic OT-I CTLs into the tumor-bearing mice, and analysis of Th1/Th2 responses by intracellular cytokine staining in CD4 and CD8 cells. RESULTS: Cryoimmunotherapy caused robust and tumor-specific CTL responses, increased Th1 responses, significantly prolonged survival and dramatically reduced lung metastasis. Although intratumor administration of dendritic cells alone increased the proliferation rate of CD8 cells, only cryoimmunotherapy resulted in the generation of effector memory cells. Furthermore, cryoimmunotherapyprotected mice that had survived primary MO5 tumors from rechallenge with parental tumors. CONCLUSIONS: These results present cryoimmunotherapy as a novel approach for systemic treatment of cancer. We envisage that cryotherapy of tumors combined with subsequent in situ immunotherapy by autologous unmodified immature dendritic cells can be applied in practice.


Subject(s)
Carcinoma, Lewis Lung/therapy , Cryotherapy/methods , Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Melanoma, Experimental/therapy , Neoplasm Metastasis/prevention & control , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Cell Proliferation/drug effects , Combined Modality Therapy , Female , Flow Cytometry , Hyaluronan Receptors/immunology , Interferon-gamma/immunology , Interleukin-4/immunology , L-Selectin/immunology , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Metastasis/immunology , Receptors, Antigen, T-Cell/genetics , Survival Analysis , Treatment Outcome
19.
J Immunol ; 174(4): 2116-23, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15699142

ABSTRACT

The magnitude of response elicited by CTL-inducing vaccines correlates with the density of MHC class I (MHC-I)-peptide complexes formed on the APC membrane. The MHC-I L chain, beta2-microglobulin (beta2m), governs complex stability. We reasoned that genetically converting beta2m into an integral membrane protein should exert a marked stabilizing effect on the resulting MHC-I molecules and enhance vaccine efficacy. In the present study, we show that expression of membranal human beta2m (hbeta2m) in mouse RMA-S cells elevates MHC-I thermal stability. RMA-S transfectants bind an exogenous peptide at concentrations 10(4)- to 10(6)-fold lower than parental RMA-S, as detected by complex-specific Abs and by T cell activation. Moreover, saturation of the transfectants' MHC-I by exogenous peptide occurs within 1 min, as compared with approximately 1 h required for parental cells. At saturation, however, level of peptide bound by modified cells is only 3- to 5-fold higher. Expression of native hbeta2m only results in marginal effect on the binding profile. Soluble beta2m has no effect on the accelerated kinetics, but the kinetics of transfectants parallel that of parental cells in the presence of Abs to hbeta2m. Ab inhibition and coimmunoprecipitation analyses suggest that both prolonged persistence of peptide-receptive H chain/beta2m heterodimers and fast heterodimer formation via lateral diffusion may contribute to stabilization. In vivo, peptide-loaded transfectants are considerably superior to parental cells in suppressing tumor growth. Our findings support the role of an allosteric mechanism in determining ternary MHC-I complex stability and propose membranal beta2m as a novel scaffold for CTL induction.


Subject(s)
Adjuvants, Immunologic/metabolism , Histocompatibility Antigens Class I/metabolism , Membrane Proteins/metabolism , beta 2-Microglobulin/metabolism , Adjuvants, Immunologic/chemical synthesis , Adjuvants, Immunologic/genetics , Animals , Binding Sites, Antibody , Binding, Competitive/immunology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Humans , Jurkat Cells , Kinetics , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/prevention & control , Membrane Proteins/chemical synthesis , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Protein Binding/immunology , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Thermodynamics , Transfection , beta 2-Microglobulin/chemical synthesis , beta 2-Microglobulin/genetics , beta 2-Microglobulin/immunology
20.
J Pept Sci ; 11(1): 37-44, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15635725

ABSTRACT

The IgG-derived immunomodulating peptide tuftsin, Thr-Lys-Pro-Arg, is recognized by specific receptors on phagocytic cells, notably macrophages, and is capable of targeting proteins and peptides to these sites. Aiming to target 3'-azido-3'-deoxythymidine (AZT) to HIV-infected macrophages, a conjugate of AZT with tuftsin was synthesized. The AZT-tuftsin chimera possesses the characteristic capacities of its two components. Thus, like AZT, it inhibits reverse transcriptase activity and HIV-antigen expression, and similarly to tuftsin, it stimulates IL-1 release from mouse macrophages and augments the immunogenic function of the cells. Importantly, the conjugate is not cytotoxic to T-cells. The results suggest that the AZT-tuftsin conjugate might have potential use in AIDS therapy.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , Macrophages/virology , Reverse Transcriptase Inhibitors/pharmacology , Tuftsin/analogs & derivatives , Tuftsin/chemical synthesis , Zidovudine/analogs & derivatives , Zidovudine/chemical synthesis , Animals , Anti-HIV Agents/toxicity , Antigen Presentation , Cell Line , Cell Survival/drug effects , Cells, Cultured , Drug Carriers , Drug Design , Female , HIV Reverse Transcriptase/metabolism , HIV-1/physiology , Humans , Interleukin-1/metabolism , Macrophages/drug effects , Macrophages/immunology , Mice , Reverse Transcriptase Inhibitors/toxicity , Tuftsin/pharmacology , Tuftsin/toxicity , Virus Replication/drug effects , Zidovudine/pharmacology , Zidovudine/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...